IMMUNOBIOLOGY

Effects of exogenous interleukin-7 on human thymus function

Yukari Okamoto, Daniel C. Douek, Richard D. McFarland, and Richard A. Koup

Immune reconstitution is a critical compo- (14 692 copies/10 000 cells) was present ments using NOD/LtSz-scid mice given nent of recovery after treatment of human in the CD1a؉CD3؊CD4؉CD8؉ stage in na- transplants of human fetal thymus and immunodeficiency virus (HIV) infection, tive thymus, suggesting that TREC gen- liver suggested that IL-7 can also directly cancer chemotherapy, and hematopoietic eration occurred following the cellular enhance TREC generation. Our results stem cell transplantation. The ability to division in this subpopulation. In a thymic provide compelling evidence that IL-7 has enhance T-cell production would benefit organ culture system, exogenous IL-7 a direct effect on increasing TCR-␣␤ rear- such treatment. We examined the effects increased the TREC frequency in fetal as rangement and indicate the potential use of exogenous interleukin-7 (IL-7) on apo- well as infant thymus, indicating in- of IL-7 for enhancing de novo naı¨ve T-cell ptosis, proliferation, and the generation creased T-cell receptor (TCR) rearrange- generation in immunocompromised pa- of T-cell receptor rearrangement excision ment. Although this increase could be tients. (Blood. 2002;99:2851-2858) circles (TRECs) in human thymus. Quanti- due to the effect of IL-7 to increase thymo- tative polymerase chain reaction demon- cyte proliferation and decrease apoptosis strated that the highest level of TRECs of immature CD3؊ cells, the in vivo experi- © 2002 by The American Society of Hematology Introduction

It has been reported that T-cell numbers are maintained in adults recombination event, which is common to approximately 70% of predominantly through the expansion of postthymic, memory thymocytes destined to become mature TCR␣␤ T cells. TRECs are T cells, whereas in infants, T cells are predominantly maintained stable and do not replicate with cellular proliferation,2,13; therefore, through the production of new naı¨ve T cells by the thymus.1 the TREC content in a peripheral cell population is proportional to However, others and we have recently demonstrated that the adult the frequency of recent thymic emigrants (RTEs). This frequency, thymus is still capable of thymopoiesis and can contribute to T-cell however, is affected not only by changes in thymic output but also reconstitution in adults.2,3 Several methods have been used to by the proliferative history of the cells. Because RTEs have TRECs measure thymopoietic capacity. Thymic size as measured by at levels that would not have been affected by peripheral expansion radiographic imaging1 and volumetric computed tomography mea- and cellular replication, quantification of TREC levels in peripheral surements4,5 have been correlated with numbers of CD4ϩCD45RAϩ blood mononuclear cells (PBMCs) represents a sensitive measure- naı¨ve T cells, and the number of phenotypically naı¨ve T cells after ment of thymopoietic capacity. transplantation has been shown to correlate with antigen-specific Although thymic function declines with age, substantial output function.6 However, there are concerns about limitations of estimat- is maintained into late adulthood.2,3 Furthermore, the adult thymus ing thymic function based on naı¨ve T-cell phenotype alone. T cells can contribute to immune reconstitution in individuals following expressing a naı¨ve phenotype are not necessarily accurate surrogate antiretroviral therapy,2,14,15 and following myeloablative chemo- markers of thymic function. Following thymic emigration, therapy and autologous hematopoietic stem cell transplantation.16 CD45RAϩ naı¨ve T cells can have a long quiescent life span,7 may T cells generated de novo from thymopoiesis have a broad TCR proliferate in an antigen-independent manner,8 or may rapidly repertoire and are theoretically more capable of responding to convert to CD45ROϩ memory/effector phenotype T cells.9 Further- neoantigens effectively.16,17 In contrast, peripheral expansion of more, naı¨ve T-cell markers may be acquired by memory T cells existing T-cell pools may lead to limited T-cell repertoires and (especially CD8ϩ T cells),9,10 further compounding the difficulty in antigen responsiveness.1,17-22 Therefore, in patients with human accurately enumerating naı¨ve T cells.11,12 immunodeficiency virus (HIV) infection or in those who have To measure thymic function more directly in humans, we received chemotherapy, the ability of the thymus to generate naı¨ve recently described an assay that quantifies an episomal DNA T cells with a broad TCR repertoire should allow for recovery of T by-product of the T-cell receptor (TCR) rearrangement process.2 cell–mediated immunity that is qualitatively better than if the These TCR rearrangement excision circles (TRECs) contain the recovery were only through expansion of pre-existing naı¨ve and signal joint sequences from the TCRAD locus ␦Rec to ␺J␣ memory T cells.

From the Vaccine Research Center, National Institute of Allergy and Infectious 02680-28-RGV (to D.C.D.). The opinions expressed are those of the author Diseases and Department of Experimental Transplantation and Immunology, (R.D.M.) and not necessarily those of the US Food and Drug Administration. Medicine Branch, National Cancer Institute, National Institutes of Health, Reprints: Richard A. Koup, Immunology Laboratory, National Institutes of Bethesda, MD; and the Center for Biologics Evaluation and Research, Food Health, Vaccine Research Center, Rm 3502, 40 Convent Dr, Bethesda, MD and Drug Administration, Rockville, MD. 20892; e-mail: [email protected]. Submitted October 1, 2001; accepted November 30, 2001. The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby Supported by a grant from Japanese Foundation of AIDS Prevention (to Y.O.), marked ‘‘advertisement’’ in accordance with 18 U.S.C. section 1734. and by a Leukemia and Lymphoma Society of America translational research grant 6540-00 and American Foundation for AIDS Research (AmFAR) grant © 2002 by The American Society of Hematology

BLOOD, 15 APRIL 2002 ⅐ VOLUME 99, NUMBER 8 2851 2852 OKAMATO et al BLOOD, 15 APRIL 2002 ⅐ VOLUME 99, NUMBER 8

Interleukin-7 (IL-7), which was originally reported as a pre-B selected and used for TOC. Each thymic fragment was cultured on cell growth factor,23 is produced by stromal cells in the thymus and polyethylene terephthalate track-etched trans-well membranes (Becton bone marrow and appears to play a role at multiple stages of T- and Dickinson, San Jose, CA) suspended in 12-well plates in UltraCULTURE B-lymphocyte development.24,25 In mice, the IL-7 receptor (IL-7R) fetal calf serum (FCS)–free medium (Biowittaker, Walkersville, MD) supplemented with 50 U/mL penicillin and 50 mg/mL streptomycin in the is first expressed in lymphoid lineage-restricted progenitors in bone presence or absence of recombinant human IL-7 (Peproteck, Rocky Hill, marrow26 and later can be detected in various tissues including NJ). The cultures were maintained in a 5% CO2 incubator at 37°C. thymus.27 IL-7R is composed of the IL-7R␣27 chain and the common cytokine receptor ␥ chain,28 the latter of which is the Treatment of NOD-SCID-hu mice with IL-7 indispensable subunit receptor for several lymphoid-related cyto- kines such as IL-2, IL-4, IL-9, and IL-15.29 Several studies have The NOD/LtSz-scid mice were purchased from Jackson Laboratory (Bar shown the essential role of this cytokine in the survival and normal Harbor, ME). Mice were implanted with pieces (ϳ2mm3 each) of human differentiation of thymocytes into mature naı¨ve T cells in mice. It fetal thymus and liver under the kidney capsule as previously described39,40 has been suggested that IL-7 is a cofactor for V(D)J rearrangement and maintained under specific pathogen-free conditions. Twenty-eight days after implantation, the size of the grafts was checked and the mice were of the TcRb locus30 and that it is also required for TcRb D-J 31 accordingly divided into 4 groups with similarly sized thymic grafts. Four rearrangement. In mice with targeted deletions of the IL-7 gene days later, 5 mice from each group were injected intraperitoneally twice Ϫ/Ϫ (IL-7 ), there was a 20-fold reduction in the number of pro–T daily with 100 ng each of IL-7 or phosphate-buffered saline. This was 32 cells, yet the percentage of TcR␣␤ thymocytes was relatively continued daily for 10 days. On days 7 and 21 after the last injection, normal,25,32 whereas the number of TcR␥␦ thymocytes was sub- IL-7–treated and control mice were autopsied and the graft, blood, spleen, stantially reduced.32 In mice with deletions of the IL-7R gene and mesenteric lymph nodes were recovered for evaluation of TREC levels (IL-7R␣Ϫ/Ϫ and ␥cϪ/Ϫ), the number of TcR␣␤ thymocytes was and phenotypic analysis. markedly decreased and TcR␥␦ thymocytes were completely absent.33,34 On the other hand, in humans, it has been reported that Flow cytometry mutations that prevent the expression of IL-7R␣ chain and ␥c result 35,36 Thymocytes were stained with combinations of the following directly in severe combined immunodeficiency (SCID). Thus, it appears labeled antihuman mouse monoclonal antibodies and isotype-matched that IL-7 is necessary for the survival and proliferation of early controls: CD1a (HI149), CD3 (SK6), CD4 (SK3), CD8 (SK1) (Becton thymic progenitor cells and production of TcR␥␦ T cells, although Dickinson); CDw127 (IL-7R␣) (R34.34) (Beckman Coulter, Brea, CA); it may not be unconditionally required for rearrangement of CD3 (UCHT1), Ki67 (B56), BrdU (3D4), annexin V, CD45RA (HI00), (BD TcRa and b in mice. However, the regulatory mechanism Pharmingen, San Diego, CA); and BCL-2 (124) (DAKO, Glostrup, of TCR␣␤ or ␥␦ rearrangement in human thymus and the roles Denmark). After staining, the cells were fixed in 1% paraformaldehyde and of IL-7 in thymopoiesis are as yet not fully understood. 4-color flow cytometry was performed using a FACSCalibur (Becton Importantly, it has been recently shown that IL-7 levels are Dickinson) flow cytometer. For Ki67 and BCL-2, the cells were first stained increased during T lymphopenia in HIV infection, strongly for surface CD3, CD4, and CD8 and then fixed and permeabilized using the suggesting that IL-7 may play a major role in both peripheral Cytofix/Cytoperm (BD Pharmingen) prior to staining of these intracellu- lar . Bromodeoxyuridine (BrdU) staining was carried out after and central T-cell homeostasis.37,38 staining for surface antigens using the BrdU Flow Kit (BD Pharmingen) We evaluated the effects of exogenous IL-7 on human thymopoi- according to the manufacturer’s instructions. The data were analyzed using esis in vitro using human thymic organ culture (TOC) and in vivo CellQuest software (Becton Dickinson). with NOD/LtSz-scid mice implanted with human thymus and liver (NOD-SCID-hu). The addition of IL-7 to TOC increased thymo- Sorting of thymocytes cyte proliferation and TREC levels and decreased apoptosis in thymuses obtained from fetuses and infants. In vivo, exogenous After mechanical disruption of thymus fragments, the cells were labeled IL-7 enhanced TREC levels in thymic grafts in NOD-SCID-hu with CD3 MicroBeads (Miltenyi Biotech, Auburn, CA) and separated into ϩ Ϫ mice. Our results indicate that IL-7 results in increased TCR CD3 and CD3 populations using separation columns (Miltenyi Biotech) according to the manufacturer’s instructions. Thymic subsets were sorted rearrangement in human thymus, both in vitro and in vivo, and by FACStar cell sorter (Becton Dickinson) after being stained for CD1a, suggest that immune reconstitution in humans could be augmented CD3, CD4, and CD8. through stimulating thymus-dependent T-cell generation with exog- enous IL-7. Quantitative polymerase chain reaction of TRECs

The sorted cells were lysed in 100 ␮g/mL proteinase K (Roche Diagnostics, Indianapolis, IN) for 3 hours at 56°C and then 20 minutes at 95°C. TREC Materials and methods levels were measured by real-time quantitative polymerase chain reaction (PCR) using the 5Ј-nuclease (TaqMan) assay in an AB17700 system Thymus and liver (Perkin-Elmer, Norwalk, CT). As previously described,16 each 25-␮L Human fetal thymus and liver tissue at 18 to 22 weeks of gestation were reaction contained 5 ␮L cell lysate, and the final concentration of each obtained from Advanced Bioscience Resources (Alameda, CA) and were component was as follows: 1.0 times reaction buffer, 20 U/mL platinum taq processed within 24 hours of harvest. Newborn, infant, and adult thymuses polymerase (Gibco BRL, Grand Island, NY), 3.5 mM MgCl2, 0.2 mM were obtained from patients undergoing corrective cardiac surgery or adult dNTPs, 500 nM each primer, 150 nM probe, and Blue-636 reference thymectomy (Children’s Medical Center or Parkland Memorial Hospital, (Megabases, Chicago, IL). The sense and antisense primers were 5Ј- Dallas, TX) and were processed within 6 hours of harvest. cacatccctttcaaccatgct-3Ј and 5Ј-cctaaaccctgcagctggc-3Ј, respectively, and the probe was FAM-acacctctggtttttgtaaaggtgcccact-TAMRA (MegaBases). TOC PCR conditions were 95°C for 5 minutes followed by 40 cycles of 95°C for 30 seconds and 60°C for 1 minute. A standard curve was plotted and TREC Thymus tissue was dissected into pieces of about 2 mm3 so that every piece values for samples were calculated using ABI7700 software. Samples were was likely to contain cortex and have similar cellularity. In adult samples, analyzed in duplicate. TRECs are present at 0, 1, or 2 copies per diploid cell only the lobes that, on visual inspection, contained cell-dense areas were and there is no pseudogene sequence. BLOOD, 15 APRIL 2002 ⅐ VOLUME 99, NUMBER 8 IL-7 AND HUMAN THYMUS 2853

Results

TREC levels in FACS-sorted thymocyte subsets in the fresh human thymus

To investigate the timing of TREC generation and dilution during thymic development in vivo, we measured TREC levels in sorted thymocyte subsets from a newborn infant. The cells were separated by magnetic cell sorting (MACS) and flow cytometry based on surface phenotype. Commitment of progenitor cells to the T-cell lineage occurs at or around the transition of CD1aϪCD34ϩ thymocytes into CD1aϩCD34ϩ thymocytes.41,42 Cells were sorted by flow cytometry for CD1aϩ/ϩϩCD3Ϫ and CD3ϩ subsets so as to exclude the CD1aϪCD3Ϫ subset, which contains precursors for natural killer (NK) cells and thymic dendritic cells.43-45 Quantita- tive PCR demonstrated that the TREC level in unfractionated thymocytes was 12 194/10 000 cells (Figure 1). No TRECs were detected at the CD3ϪCD4ϪCD8Ϫ triple negative (TN), CD3ϪCD4lowCD8Ϫ,orCD3ϪCD4ϩCD8Ϫ stages of thymocyte Figure 2. IL-7R␣ (CD127) expression in thymuses from subjects of varying ages. Freshly isolated thymocytes from a 22-week gestation fetus, a 2-month-old development. However, the highest level of TRECs was detected infant, and a 14-year-old youngster were analyzed by flow cytometry for expression within CD1aϩ cells that are CD3ϪCD4ϩCD8ϩ double positive of IL-7R␣ (CD127) on multiple cell types. (CD3Ϫ DP; 14 692/10 000 cells), confirming previous experiments that address the timing of TCRD gene rearrangement.46 This indicates that the excision leading to TRECs that commits a cell to vary depending on the possible TCR deletion on only one allele in the TCR␣␤ lineage occurs concomitant with the expansion of part of the cells.48 Because cellular divisions after TREC genera- CD3Ϫ DP thymocytes. If we assume that TCRD deletion occurs in tion will dilute TRECs, these results indicate that the initiation of both alleles,47 then a TREC level of 14 692/10 000 cells would cell expansion precedes TREC generation in CD3Ϫ DP cells. High indicate than an average of 75% of cells have undergone rearrange- levels of TREC were still detected in the CD3ϩ DP subset, which ment of both alleles to generate TREC. However, this value may had undergone expansion (6900/10 000 cells), and also in the more mature CD4ϩ and CD8ϩ single positive (SP) subsets (6064 and 4099/10 000 cells, respectively). The decrease in TRECs in mature cells is likely to represent dilution secondary to cellular prolifera- tion that occurs during the processes of positive and negative selection. There was no evidence to suggest that TREC generation still occurs after the CD3ϩ stage.

IL-7R␣ expression in thymocyte subsets

As a basis for studying the role of IL-7 in TREC generation in human thymus, IL-7R␣ expression was examined by flow cytom- etry on thymocytes from a fetus (22 weeks’ gestation), a 2-month- old infant, and a 14-year-old youth (Figure 2). IL-7R␣ expression was highest in immature CD3ϪCD4lowCD8Ϫ cells (89.0%-95.5%), just before TREC generation (Figures 1 and 2). Because cells with this phenotype are thought to be initiating the process of TCR␣␤ or ␥␦ lineage commitment and are known to undergo proliferation,49 these results suggest the involvement of IL-7R␣ in regulating the proliferation of thymocytes and their commitment to TCR␣␤ or ␥␦ lineage. Although IL-7R␣ expression tended to decrease after the Figure 1. TREC levels in FACS-purified thymocyte subsets from the human Ϫ low Ϫ thymus. TREC levels were measured in FACS-sorted cells from a fresh thymus of a CD3 CD4 CD8 stage, it was still fairly high (52.2%-85.9%) in newborn infant. For CD3Ϫ cells, the thymocytes were first depleted of CD3ϩ cells by CD3Ϫ DP cells regardless of the age of the thymus. IL-7R␣ MACS and then sorted by FACS for the subsets based on CD4 and CD8 expression expression was lowest in CD3ϩ DP cells (31.4%-57.7%) and then (dot plot, upper right) to more precisely determine the timing of peak TREC ϩ production. Cells were also sorted for CD1aϩ/ϩϩ cells so as to exclude the increased again in CD3 SP cells (43.6%-69.2%). CD1aϪCD3Ϫ subset, which may contain precursors for NK cells and thymic dendritic cells. The sorted cells were subjected to TREC assay. The CD3ϩ-depleted fraction The effects of IL-7 on proliferation and apoptosis (shown as R4 in the histogram, upper left) contained cells expressing CD3 at the of thymocytes intermediate level (referred to as CD3low) as well as CD3Ϫ cells. On the other hand, Ϫ Ϫ/low for measuring the frequency of each subpopulation in CD3 and CD3 popula- Several studies have suggested that IL-7 is a physiologic survival tions, we analyzed whole thymocytes from the same thymus sample using gate R1. 24,50-52 For CD3ϩ subsets, the whole thymocytes were analyzed for frequency measurement factor for early lymphoid progenitor cells. The evaluation of using gates R2 or R3 and then sorted for the TREC assay. Therefore, it should be TREC levels in each thymic subpopulation will be affected by noted that the actual frequency of CD3Ϫ/low CD4ϩCD8ϩ and CD3ϩCD4ϩCD8ϩ cells proliferation and apoptosis of thymocytes. To evaluate the in vitro should be slightly higher and lower, respectively, than the values shown here (36.4% and 35.7%, respectively). The TREC values are expressed as copies per 10 000 effects of exogenous IL-7 on TREC generation in our TOC system, cells. These data represent 3 different experiments. we first assessed the effect of IL-7 on thymocyte proliferation by 2854 OKAMATO et al BLOOD, 15 APRIL 2002 ⅐ VOLUME 99, NUMBER 8

respectively) but also in more mature CD3ϩCD4ϩ SP (6.6 Ϯ 3.3- fold) and CD3ϩCD8ϩ SP cells (18.8 Ϯ 8.6-fold). This effect was dependent on IL-7 dose (data not shown). We next assessed the effects of IL-7 on apoptosis in TOCs by measuring expression of annexin V after 4 days of TOC in the presence or absence of IL-7. IL-7 significantly lowered annexin V expression by about 50% in CD3ϪCD8Ϫ immature thymocytes (Figure 3B, upper and lower panels), which were pre–TREC- generating cells (Figure 1). We then examined BCL-2, a well- established , which is involved in the suppression of apoptosis. It has been reported that BCL-2 expression during T-cell differentiation is multiphasic; it is high in CD3Ϫ immature thymocytes, down-regulated in CD4ϩCD8ϩ thymocytes to facili- tate selection process, and up-regulated in mature CD3ϩ SP thymocytes thus enhancing prolonged survival.53 As expected, IL-7 enhanced BCL-2 expression in CD3Ϫ immature thymocytes in a dose-dependent way (Figure 3C). These results were consistent with previous studies in mice demonstrating that short-term culture with IL-7 of immature thymocytes from IL-7Ϫ/Ϫ mice caused up-regulation of BCL-2 and increased cell survival.51 Thus, in human thymus, IL-7 led to an increase in proliferation and decrease in apoptosis of immature CD3Ϫ to CD3low cells, which correlated with the increased expression of BCL-2.

The in vitro effects of IL-7 on TREC generation in TOCs

The effects of IL-7 on TREC generation were examined in TOCs. After culturing newborn thymus in the presence (10 and 50 ng/mL) or absence of IL-7 for 4 days, thymocytes were separated into CD3Ϫ and CD3ϩ fractions by MACS (Figure 4A). The majority of CD3low cells were contained in the CD3Ϫ fraction (Figure 4A), whereas most of CD3ϩ DP cells were collected into the CD3ϩ fraction (data not shown). Figure 4B shows the TREC frequency in

Figure 3. Proliferation and apoptosis of thymocytes in response to IL-7. (A) The incorporation of BrdU was measured in thymocytes in the presence (10 ng/mL) or absence of IL-7 on day 4 of TOC. Upper panel shows representative results from 5 different experiments. Ratio of IL-7 to control is shown in the lower panel. (B) The inhibition effect of IL-7 on apoptosis of thymocytes was evaluated by flow cytometry by the expression of annexin V after 4 days of TOC. Upper panel shows the representative result from 5 different experiments. Lower panel shows the percent inhibition of annexin V expression by IL-7 to control TOC. BCL-2 expression was evaluated in TOC with 1, 10, and 50 ng/mL IL-7. Data are expressed as mean value Ϯ SD in triplicate. measuring the incorporation of BrdU. The upper panel of Figure 3A represents several experiments using different thymuses. The Figure 4. The in vitro effects of IL-7 on TREC generation in TOCs. Thymocytes rate of BrdU incorporation was highest in CD3Ϫ/lowCD4ϩCD8Ϫ from a newborn thymus were separated by MACS for CD3Ϫ and CD3ϩ subsets cells and low in mature CD3ϩ cells (Figure 3A). We observed a before or after 4 days of TOC in the absence or presence of IL-7 (10 and 50 ng/mL) and TREC levels were measured. (A) The representative data of CD3 expression is marked increase in BrdU uptake in most thymocyte populations shown in CD3Ϫ (thin line) and CD3ϩ (bold line) fractions sorted by selection column with the administration of 10 ng/mL IL-7 (Figure 3A). Increased from preculture and day 4 of TOC in the absence or presence of IL-7 (10 ng/mL). (B) BrdU uptake with IL-7 was observed consistently in 5 different TREC levels in the 2 fractions were measured. Three separate experiments were performed. Each experiment was performed in quadruplicate with all 3 concentra- TOCs (Figure 3A, lower panel), not only in immature cells tions of IL-7. A representative result is shown with error bars pertaining to the (4.0 Ϯ 3.3-fold and 2.3 Ϯ 0.8-fold in TN and CD3ϪCD4lowCD8Ϫ, experiment. BLOOD, 15 APRIL 2002 ⅐ VOLUME 99, NUMBER 8 IL-7 AND HUMAN THYMUS 2855

TOCs. IL-7 increased TREC levels in both CD3Ϫ and CD3ϩ checking the size of the grafts. TREC levels in thymocytes from the subsets in a dose-dependent way. Although there was a difference grafts were significantly higher in the IL-7–treated group (14 276 Ϯ between 10 ng/mL IL-7–treated and untreated TOC, the only 1290 and 14 344 Ϯ 2449 for day 7 and day 21, respectively) than statistically significant difference existed between the 50 ng/mL the control group (8135 Ϯ 1070 and 9877 Ϯ 1869 for day 7 and day and untreated TOCs (P Ͻ .09 and P Ͻ .001 in CD3Ϫ and CD3ϩ 21, respectively) at day 7 (P Ͻ .002) and day 21 (P Ͻ .007) after subsets, respectively). the last administration of IL-7 (Figure 5B). Although the average If IL-7 were to be used as a therapeutic agent to improve graft weight (Figure 5C) was higher in IL-7–treated mice after 7 immune reconstitution, its effect would have to be maintained in days, the difference between the 2 groups was not statistically postnatal thymuses. The effects of IL-7 on TREC levels were significant either after 7 or 21 days. Moreover, there was no change further evaluated in TOCs using thymuses from subjects of by IL-7 treatment in the percentage of Ki67ϩ cells in any of the differing ages. Fetal (18, 19, and 22 weeks’ gestation), newborn (2 subsets except for CD3ϩ DP and CD3ϩ CD8 SP at day 7 (Figure and 15 day, and 3, 4, 7, and 11 month), and infant (3 and 5.6 year) 5D). Also, there was no significant increase either in the percentage thymuses were placed in TOCs in the presence of IL-7, and TRECs of human CD3ϩ cells or TREC levels in the peripheral blood after were measured (Table 1). In all TOCs, TREC levels increased in the IL-7 treatment (data not shown). presence of IL-7 (1.3 Ϯ 0.1-fold for whole cells, 2.0 Ϯ 0.8-fold for CD3Ϫ fractions, and 1.5 Ϯ 0.2-fold for CD3ϩ fractions).

The in vivo effects of IL-7 on TREC generation Discussion in NOD-SCID-hu mice It is well established that IL-7 exerts potent effects on T-cell Several studies have demonstrated the in vivo effects of IL-7 on progenitors and is required for T-cell development. It is critical for mouse thymopoiesis.54,55 In the present study we used NOD- homeostatic proliferation and survival of not only thymocytes but SCID-hu chimeric mice to measure the in vivo effects of IL-7 on also naı¨ve T cells in the peripheral blood and lymph nodes.56 In the human thymopoiesis. Twenty-eight days after engraftment of present study we used the quantitative TREC assay to assess more human fetal thymus and liver under the mouse kidney capsule, the definitively the effects of exogenous IL-7 on TCR rearrangement in growth of thymic grafts was readily apparent; typically, a 2-mm3 thymocytes using both in vitro TOCs and thymic engraftments in fragment grew up to a diameter of 5 to 7 mm. These grafts NOD-SCID mice. contained comparable levels of TRECs per thymocytes to those of To understand the timing of TREC generation and its dilution native human thymuses (data not shown), indicating the generation during thymic development, we quantified TREC level in each of new thymocytes in the grafts. More importantly, we were able to thymic subpopulation. We found the highest level of TREC in the detect human CD45ϩ cells in the peripheral blood and spleen. Most CD1aϩCD3Ϫ DP stage. Recently, TCR␤ selection in the human of these human cells were positive for CD3 (data not shown) and thymus was reported to be initiated at the transition of represented 1.0% and 1.3% of the total lymphocytes in PBMCs and CD3ϪCD4ϩCD8Ϫ into the CD4ϩCD8␣ϩ␤Ϫ stage,57 whereas TCRA spleen, respectively (Figure 5A, upper panel). These CD3ϩ cells has been known to be restricted to CD3ϩ DP and comprised mature CD4ϩ and CD8ϩ SP cells (Figure 5A, middle later stages of T-cell development. Together with these, our data panel), the majority of which were CD45RAϩ, suggesting that they suggest that TCRB gene rearrangement precedes and induces the were naı¨ve T cells (Figure 5A, bottom panel). The frequency of initiation of TREC generation, followed by TCRA rearrangement. human CD3ϩCD45RAϩCD4ϩ or CD3ϩCD45RAϩCD8ϩ T cells TREC generation may represent a decisive step in the ␣␤ versus ␥␦ correlated positively with the size of the graft (data not shown). lineage commitment of differentiating thymocytes because this is We treated NOD-SCID-hu mice with 100 ng IL-7 (n ϭ 11) or an intermediate rearrangement between TCRD and TCRA gene saline (n ϭ 8) twice daily for 10 days beginning 4 days after rearrangement, which excises most of the TCRD locus.58 The

Table 1. The effect of IL-7 on TREC levels in TOCs using thymuses of differing ages TREC/10,000 cells Day 4 or Day 5* Whole CD3Ϫ CD3ϩ TOC no. Age of thymus Control IL-7 Ratio† Control IL-7 Ratio† Control IL-7 Ratio†

1 Fetal 18 wk-1 4 738 6 074 1.3 4 120 13 561 3.3 ND ND ND 2 18 wk-2 ND ND ND 4 047 11 318 2.8 ND ND ND 3 19 wk ND ND ND 3 970 4 883 1.2 6 618 8 347 1.3 4 22 wk ND ND ND 1 703 3 885 2.3 3 310 5 321 1.6 5 Newborn 2 d 10 639 15 265 1.4 7 733 13 462 1.7 7 808 12 697 1.6 6 15 d 9 790 12 333 1.3 2 080 5 044 2.4 7 507 9 751 1.3 7 3 mo 4 682 6 943 1.5 ND ND ND ND ND ND 8 4 mo 5 175 6 756 1.3 2 616 3 232 1.2 ND ND ND 9 7 mo 6 071 7 503 1.2 2 389 5 585 2.3 9 700 14 979 1.5 10 11 mo 5 820 7 884 1.4 4 195 4 387 1.0 5 768 8 416 1.5 11 Infant 3 y 8 590 12 183 1.4 ND ND ND ND ND ND 12 5.6 y 8 297 10 200 1.2 ND ND ND ND ND ND Mean Ϯ SD 1.3 Ϯ 0.1 2.0 Ϯ 0.8 1.5 Ϯ 0.2

ND indicates not done. *TOC no. 3 and 11 were cultured for 5 days and others were cultured for 4 days. †IL-7(ϩ)/IL-7(Ϫ). 2856 OKAMATO et al BLOOD, 15 APRIL 2002 ⅐ VOLUME 99, NUMBER 8

Figure 5. The in vivo effects of IL-7 on TREC generation in NOD-SCID-hu mice. Four weeks after implantation of human fetal thymus and liver, NOD-SCID-hu mice were treated with 100 ng IL-7 twice daily for 10 days. Seven (control, n ϭ 3; IL-7, n ϭ 4) and 21 days (control, n ϭ 5; IL-7, n ϭ 7) after the last injection, the grafts, spleen, and PBMCs were recovered from the mice for phenotypic analysis and TRECs. The upper panels of panel A indicate percentage of human CD3ϩ cells and the panels in the middle row of panel A indicate human CD4ϩ and CD8ϩ expression on the CD3ϩ population of PBMC (left) and spleen (right). The bottom figures of panel A show CD45RAϩ cells on CD3ϩCD4ϩ or CD3ϩCD8ϩ populations of PBMCs (left) and spleen (right). The TREC levels of grafts and the graft weights in control and IL-7–treated mice are shown in panels B and C, respec- tively. The percentages of Ki67ϩ cells in each subpopulation of grafts are shown in panel D.

only apparent reason for this rearrangement is to delete the expression in any of the subsets did not show a significant change TCRD locus to prepare the allele for subsequent TCRA gene by IL-7 treatment (Figure 5C and 5D, respectively). Considering rearrangement. Thus, our finding that IL-7 increases TREC that IL-7 can exhibit distinct effects on T cells at different levels of thymocytes most likely indicates that IL-7 eventually concentrations,60 one possible interpretation of our data would be leads to the enhancement of induction of TCRA and TCRB gene that the endogenous human IL-7 in these grafts was sufficient to rearrangement. maintain the normal development of thymocytes, whereas the Although IL-7 increased TREC in our TOC system, we cannot addition of exogenous IL-7 further promoted TCR gene rearrange- definitively prove that this is due to the promotion of TREC ment and therefore TREC generation, but was not enough to generation in the individual thymocytes because TREC levels are promote further proliferation of immature cells. In this experiment also affected by the ability of IL-7 to regulate apoptosis and we did not measure the rate of apoptosis in the thymic grafts. proliferation of cells at the pre–TREC-generating stage.49,50,51,59 However, it is unlikely that this increase in TREC by IL-7 was Indeed, BrdU, annexin V, and BCL-2 staining in our TOC system caused solely by lengthening the survival of immature cells clearly demonstrated that exogenous IL-7 can enhance the prolifera- because the percentage of annexin Vϩ cells in fresh human thymus tion and prevent apoptosis of immature CD3Ϫ/low subsets (Figure 3) is typically as low as 0.5% to 4.0% in each subpopulation (Yukari which, according to our results (Figure 1), precede TREC genera- Okamoto unpublished data, May 1999). Therefore, the logical tion. It is quite possible, therefore, that IL-7 indirectly increases the interpretation is that IL-7 can directly stimulate TREC generation. TREC frequency in the thymus by promoting the proliferation and Based on transgenic mouse studies, it has been suggested that there survival of thymocytes pre-TREC generation. is a regulatory sequence near the human ␦Rec gene segment, which On the other hand, our in vivo experiments suggested another is recognized by putative regulatory proteins that induce ␦Rec pathway of IL-7 effects. Exogenous IL-7 was administered to rearrangement.61 Moreover, transcription of the T-early ␣ (TEA) NOD-SCID-hu mice, where the growth of human thymic grafts element is assumed to be responsible for opening the TCRAJ locus appeared to be fairly active. At 7 and 21 days after the last to the V(D)J recombinase complex.62,63 Further identification of administration, TREC levels in thymocytes from the grafts were regulatory mechanism of the ␦Rec-␺J␣ recombination event will significantly higher in the IL-7–treated group than the control eventually allow a determination of whether IL-7 has a direct effect group (Figure 5B). However, the graft weights and also Ki67 on intracellular processes that lead to enhanced TREC production. BLOOD, 15 APRIL 2002 ⅐ VOLUME 99, NUMBER 8 IL-7 AND HUMAN THYMUS 2857

In our NOD-SCID-hu mouse system, we observed no signifi- practice it may be necessary to antagonize the effects of thymic cant increase for the percentage of human CD3ϩ cells in the atrophic factors.70 The decrease in thymocyte apoptosis mediated peripheral blood as a result of IL-7 treatment. It has been reported by high concentrations of exogenous IL-7 in CD3ϩ DP population that mouse NK cells affect the efficiency of engraftment of human may affect negative selection. Furthermore, it should also be cells in this model.64 Although NOD/LtSz-scid mice have rela- considered that high concentrations of exogenous IL-7 may break tively low NK cell activity,65 it is possible that IL-7 caused an unresponsiveness of residual autoreactive T cells in healthy individu- enhancement of this.66,67 Be that as it may, our data clearly show als by stimulating the anergic T cells that suppress the autoreactive that IL-7 increased TCR gene rearrangement and TREC genera- cells71 and may cause autoimmune disease. Nevertheless, our tion, which would suggest that thymopoietic function was aug- results provide a basis for understanding some of the regulatory mented in vivo. mechanisms of thymocyte development in the human thymus and Complete recovery of broad T-cell immunity after insults such encourage the further evaluation of cytokine-based immune recon- as HIV infection or chemotherapy will require the generation of stitution strategies, through the stimulation of thymus-dependent new naı¨ve T cells from the thymus.2,16,17,21 The administration of T-cell generation. thymopoietic cytokines may be able to improve immune reconstitu- tion by augmenting thymic function in these situations. IL-7 is a major factor for thymopoiesis, and systemic administration of recombinant IL-7 to murine bone marrow transplant recipients has Acknowledgments been shown to normalize thymopoiesis and improve immune function after bone marrow transplantation.21,68,69 Furthermore, We thank Brenna Hill, Joseph Beckham, and Angie Mobley for endogenous IL-7 may play a major role in the homeostatic response technical help; Dr Michael Betts and Dr Takeshi Kurata for helpful to T lymphopenia.37 Our data suggest that IL-7 can enhance the advice; and Dr Steven Leonard for procurement of postnatal function of fetal and infant thymuses, although in therapeutic thymuses. References

1. Mackall CL, Fleisher TA, Brown MR, et al. Age, 12. Young JL, Ramage JM, Gaston JS, Beverley PC. 23. Namen AE, Schmierer AE, March CJ, et al. B cell thymopoiesis, and CD4ϩ T-lymphocyte regenera- In vitro responses of human CD45R0brightRAϪ precursor growth-promoting activity: purification tion after intensive chemotherapy. N Engl J Med. and CD45R0-RAbright T cell subsets and their rela- and characterization of a growth factor active on 1995;332:143-149. tionship to memory and naive T cells. Eur J Im- lymphocyte precursors. J Exp Med. 1988;167: 2. Douek CD, McFarland DR, Keiser HP, et al. munol. 1997;27:2383-2390. 988-1002. Changes in thymic function with age and during 13. Kong F, Chen CH, Cooper MD. Thymic function 24. Peschon JJ, Morryssey PJ, Grabstein KH, et al. the treatment of HIV infection. Nature. 1998;396: can be accurately monitored by the level of recent Early lymphocyte expansion is severely impaired 690-695. T cell emigrants in the circulation. Immunity. in interleukin 7 receptor-deficient mice. J Exp 3. Jamieson BD, Douek DC, Killian S, et al. Genera- 1998;8:97-104. Med. 1994;180:1955-1960. tion of functional thymocytes in the human adult. 14. Poulin JF, Sekaly RP. Function of the thymus in 25. Von Freeden-Jeffry U, Vieira P, Lucian LA, McNeil Immunity. 1999;10:569-575. HIV-infected adults. JAMA. 1999;282:219. T, Burdach S, Murry R. Lymphopenia in interleu- 4. McCune JM, Loftus R, Schmidt DK, et al. High 15. Zhang L, Lewin SR, Markowitz M, et al. Measur- kin (IL)-7 gene-depleted mice identifies IL-7 as a prevalence of thymic tissue in adults with human ing recent thymic emigrants in blood of normal nonredundant cytokine. J Exp Med. 1995;181: immunodeficiency virus-1 infection. J Clin Invest. and HIV-1-infected individuals before and after 1519-1526. effective therapy. J Exp Med. 1999;190:725-732. 1998;101:2301-2308. 26. Kondo M, Takeshita T, Higuchi M, et al. Func- 5. Smith KY, Valdez H, Landay A, et al. Thymic size 16. Douek DC, Vescio RA, Betts MR, et al. Assess- tional participation of the IL-2 receptor gamma and lymphocyte restoration in patients with hu- ment of thymic output in adults after haematopoi- chain in IL-7 receptor complexes. Science. 1994; man immunodeficiency virus infection after 48 etic stem-cell transplantation and prediction of 263:1453-1454. weeks of zidovudine, lamivudine, and ritonavir T-cell reconstitution. Lancet. 2000;355:1875- 1881. 27. Goodwin RG, Friend D, Ziegler SF, et al. Cloning therapy. J Infect Dis. 2000;181:141-147. of the human and murine interleukin-7 receptors: 17. Heitger A, Neu N, Kern H, et al. Essential role of 6. Socie G, Stone JV, Wingard JR, et al. Long-term demonstration of a soluble form and homology the thymus to reconstitute naive (CD45RAϩ)T- survival and late deaths after allogeneic bone to a new receptor superfamily. Cell. 1990;60: helper cells after human allogeneic bone marrow marrow transplantation. Late Effects Working 941-951. transplantation. Blood. 1997;90:850-857. Committee of the International Bone Marrow 18. Keever CA, Small TN, Flomenberg N, et al. Im- 28. Noguchi M, Nakamura Y, Russell SM, et al. Inter- Transplant Registry. N Engl J Med. 1999;341: leukin-2 receptor gamma chain: a functional com- 14-21. mune reconstitution following bone marrow trans- plantation: comparison of recipients of T-cell de- ponent of the interleukin-7 receptor. Science. 7. McLean AR, Michie CA. In vivo estimates of divi- pleted marrow with recipients of conventional 1993;262:1877-1880. sion and death rates of human T lymphocytes. marrow grafts. Blood. 1989;73:1340-1350. 29. Lin JX, Migone TS, Tsang M, et al. The role of Proc Natl Acad Sci U S A. 1995;92:3707-3711. 19. Mackall CL, Bare CV, Granger LA, Sharrow SO, shared receptor motifs and common Stat proteins 8. Soares MV, Borthwick NJ, Maini MK, et al. IL-7- Titus JA, Gress RE. Thymic-independent T cell in the generation of cytokine pleiotropy and re- ϩ dependent extrathymic expansion of CD45RA regeneration occurs via antigen-driven expansion dundancy by IL-2, IL-4, IL-7, IL-13, and IL-15. Im- T cells enables preservation of a naive repertoire. of peripheral T cells resulting in a repertoire that munity. 1995;2:331-339. J Immunol. 1998;161:5909-5917. is limited in diversity and prone to skewing. J Im- 30. Muegge K, Vila MP, Durum SK. Interleukin-7: a 9. Picker LJ, Treer JR, Ferguson-Darnell B, Collins munol. 1996;156:4609-4616. cofactor for V(D)J rearrangement of the T cell re- PA, Buck D, Terstappen LW. Control of lympho- 20. Storek J, Witherspoon RP, Storb R. T cell recon- ceptor beta gene. Science. 1993;261:93-95. cyte recirculation in man, I: differential regulation stitution after bone marrow transplantation into 31. Tsuda S, Reike S, Hashimoto Y, Nakauchi H, Ta- of the peripheral lymph node homing receptor adult patients does not resemble T cell develop- kahama Y. IL-7 supports D-J but not V-DJ rear- L-selection on T cells during the virgin to memory ment in early life. Bone Marrow Transplant. 1995; rangement of TCR-␤ gene in fetal liver progenitor cell transition. J Immunol. 1993;150:1105-1121. 16:413-425. cells. J Immunol. 1996;156:3233-3242. 10. Wills MR, Carmichael AJ, Weekes MP, et al. Hu- 21. Weinberg K, Annett G, Kashyap A, Lenarsky C, 32. Moore TA, von Freeden-Jeffry U, Murray R, Zlot- man virus-specific CD8ϩ CTL clones revert Forman SJ, Parkman R. The effect of thymic nik A. Inhibition of gamma delta T cell develop- from CD45ROhigh to CD45RAhigh in vivo: function on immunocompetence following bone ment and early thymocyte maturation in IL-7 Ϫ/Ϫ CD45RAhighCD8ϩ T cells comprise both naive marrow transplantation. Biol Blood Marrow Trans- mice. J Immunol. 1996;157:2366-2373. and memory cells. J Immunol. 1999;162:7080- plant. 1995;1:18-23. 7087. 22. Small TN, Papadopoulos EB, Boulad F, et al. 33. Maki K, Sunaga S, Komagata Y, et al. Interleukin 11. Hamann D, Baars PA, Rep MH, et al. Phenotypic Comparison of immune reconstitution after unre- 7 receptor-deficient mice lack ␥␦ T cells. Proc and functional separation of memory and effector lated and related T-cell-depleted bone marrow Natl Acad Sci. U S A. 1996;93:7172-7177. human CD8ϩ T cells. J Exp Med. 1997;186:1407- transplantation: effect of patient age and donor 34. He YW, Malek TR. Interleukin-7 receptor alpha is 1418. leukocyte infusions. Blood. 1999;93:467-480. essential for the development of gamma delta ϩ 2858 OKAMATO et al BLOOD, 15 APRIL 2002 ⅐ VOLUME 99, NUMBER 8

T cells, but not natural killer cells. J Exp Med. deletion during human T cell differentiation. Int growth factor in early human T-cell development. 1996;184:289-293. Immunol. 1998;10:1873-1880. Blood. 1996;88:4239-4245. 35. Puel A, Ziegler SF, Buckley RH, Leonard WJ. De- 47. Petrie H, Livak F, Burtrum D, et al. T cell receptor 60. Offner F, Plum J. The role of interleukin-7 in early fective IL7R expression in T(Ϫ)B(ϩ)NK(ϩ) se- gene recombination patterns and mechanisms: T-cell development. Leuk Lymphoma. 1998;30: vere combined immunodeficiency. Nat Genet. cell death, rescue, and T cell production. J Exp 87-99. 1998;20:394-397. Med. 1995;182:121-127. 61. Shutter J, Cain JA, Ledbetter S, Rogers MD, 36. Noguchi M, Yi H, Rosenblatt HM, et al. Interleu- 48. van Dongen JJ, Wolvers-Tettero IL. Analysis of Hockett RD. A ␦ T-cell receptor deleting element kin-2 receptor gamma chain mutation results in immunoglobulin and T cell receptor genes. Part II: transgenic reporter construct is rearranged in ␣␤ X-linked severe combined immunodeficiency in possibilities and limitations in the diagnosis and but not ␥␦ T-cell lineages. Mol Cell Biol. 1995;15: humans. Cell. 1993;73:147-157. management of lymphoproliferative diseases and 7022-7031. related disorders. Clin Chim Acta. 1991;198:93- 62. de Chasseval R, de Villartay JP. Functional char- 37. Napolitano LA, Grant RM, Deeks SG, et al. In- 174. acterization of the promoter for the human germ- creased production of IL-7 accompanies HIV-1- line T cell receptor J alpha (TEA) transcript. Eur mediated T-cell depletion: implications for T-cell 49. Wu L, Scollay R, Egerton M, Peares M, Span- J Immunol. 1993;23:1294-1298. homeostasis. Nat Med. 2001;7:73-79. grude GJ, Shortman K. CD4 expressed on earli- est T-lineage precursor cells in the adult murine 63. Sleckman BP, Bardon CG, Ferrini R, Davidson L, 38. Fry TJ, Christensen BL, Komschlies KL, Gress thymus. Nature. 1991;349:71-74. Alt FW. Function of the TCR alpha enhancer in RE, Mackall CL. Interleukin-7 restores immunity alphabeta and gammadelta T cells. Immunity. 50. Murray R, Suda T, Wrighton N, Lee F, Zlotnik A. in athymic T-cell-depleted hosts. Blood. 2001;97: 1997;7:505-515. 1525-1533. IL-7 is a growth and maintenance factor for ma- ture and immature thymocyte subsets. Int Immu- 64. Murphy WJ, Kumar V, Bennett M. Rejection of 39. McCune JM, Namikawa R, Kaneshima H, Shultz nol. 1989;1:526-531. bone marrow allografts by mice with severe com- LD, Lieberman M, Weissman IL. The SCID-hu bined immune deficiency (SCID): evidence that mouse: murine model for the analysis of human 51. Moore TA, Zlotnik A. T-cell lineage commitment natural killer cells can mediate the specificity of hematolymphoid differentiation and function. Sci- and cytokine responses of thymic progenitors. marrow graft rejection. J Exp Med. 1987;165: ence. 1988;241:1632-1639. Blood. 1995;86:1850-1860. 1212-1217. 40. Okamoto Y, Eda Y, Ogura A, et al. In SCID-hu 52. von Freeden-Jeffry U, Solvason N, Howard M, 65. Shultz LD, Schweitzer PA, Christianson SW, et al. mice, passive transfer of a humanized antibody Murray R. The earliest T lineage-committed cells Multiple defects in innate and adaptive immuno- prevents infection and atrophic change of me- depend on IL-7 for Bcl-2 expression and normal logic function in NOD/LtSz-scid mice. J Immunol. dulla in human thymic implant due to intravenous cell cycle progression. Immunity. 1997;7:147-154. 1995;154:180-191. inoculation of primary HIV-1 isolate. J Immunol. 53. Gratiot-Deans J, Merino R, Nunez G, Turka LA. 66. Dadmarz R, Bockstoce DC, Golub SH. Interleu- 1998;160:69-76. Bcl-2 expression during T-cell development: early kin-7 selectively enhances natural kill cytotoxicity 41. Blom B, Res P, Noteboom E, Weijer K, Spits H. loss and late return occur at specific stages of mediated by the CD56bright natural killer subpopu- Prethymic CD34ϩ progenitors capable of devel- commitment to differentiation and survival. Proc lation. Lymphokine Cytokine Res. 1994;13:349- oping into T cells are not committed to the T cell Natl Acad Sci U S A. 1994;91:10685-10689. 357. lineage. J Immunol. 1997;158:3571-3577. 54. Aspinall R, Andrew D. Thymic atrophy in the 67. Wong EK, Eaves C, Klingemann HG. Compari- 42. Spits H, Blom B, Jaleco AC, et al. Early stages in mouse is a soluble problem of the thymic environ- son of natural killer activity of human bone mar- the development of human T, natural killer and ment. Vaccine. 2000;18:1629-1637. row and blood cells in cultures containing IL-2, IL-7 and IL-12. Bone Marrow Transplant. 1996; thymic dendritic cells. Immunol Rev. 1998;165: 55. Abdul-Hai A, Or R, Slavin S, et al. Stimulation of 18:63-71. 75-86. immune reconstitution by interleukin-7 after syn- 68. Bolotin E, Smogorzewska M, Smith S, Widmer M, 43. Marquez C, Trigueros C, Fernandez E, Toribio geneic bone marrow transplantation in mice. Exp Weinberg K. Enhancement of thymopoiesis after ML. The development of T and non-T cell lin- Hematol. 1996;24:1416-1422. bone marrow transplant by in vivo interleukin-7. eages from CD34ϩ human thymic precursors can 56. Tan JT, Dudl E, LeRoy E, et al. IL-7 is critical for Blood. 1996;88:1887-1894. be traced by the differential expression of CD44. homeostatic proliferation and survival of naive T J Exp Med. 1995;181:475-483. cells. Proc Natl Acad Sci U S A. 2001;98:8732- 69. Bolotin E, Annett G, Parkman R, Weinberg K. Se- 8737. rum levels of IL-7 in bone marrow transplant re- 44. Res P, Martinez-Caceres E, Cristina Jaleco A, et cipients: relationship to clinical characteristics ϩ dim al. CD34 CD38 cells in the human thymus can 57. Blom B, Verschuren MC, Heemskerk MH, et al. and lymphocyte count. Bone Marrow Transplant. differentiate into T, natural killer, and dendritic TCR gene rearrangements and expression of the 1999;23:783-788. cells but are distinct from pluripotent stem cells. pre-T cell receptor complex during human T-cell 70. Sempowski GD, Hale LP, Sundy JS, et al. Leuke- Blood. 1996;87:5196-5206. differentiation. Blood. 1999;93:3033-3043. mia inhibitory factor, oncostatin M, IL-6, and stem 45. Dalloul AH, Patry C, Salamero J, Canque B, 58. Hockett RD, de Villartay JP, Pollock K, Poplack cell factor mRNA expression in human thymus Grassi F, Schmitt C. Functional and phenotypic DG, Cohen DI, Korseyer SJ. Human T-cell anti- increases with age and is associated with thymic analysis of thymic CD34ϩCD1aϪ progenitor- gen receptor (TCR) delta-chain locus and ele- atrophy. J Immunol. 2000;164:2180-2187. ϩ derived dendritic cells: predominance of CD1a ments responsible for its deletion are within the 71. Filion MC, Bradley AJ, Devine DV, Decary F, differentiation pathway. J Immunol. 1999;162: TCR alpha-chain locus. Proc Natl Acad Sci U S A. Chartrand P. Autoreactive T cells in healthy indi- 5821-5828. 1988;85:9694-9698. viduals show tolerance in vitro with characteris- 46. Verschuren MC, Blom B, Bogers AJ, Spits H, ven 59. Plum J, De Smedt M, Leclercq G, Verhasselt B, tics similar to but distinct from clonal anergy. Eur Dongen JJ. PJA-BP expression and TCR delta Vandekerckhove B. Interleukin-7 is a critical J Immunol. 1995;25:3123-3127.